151
|
Rangarajan N, Gordy CL, Askew L, Bevill SM, Elston TC, Errede B, Hurst JH, Kelley JB, Sheetz JB, Suzuki SK, Valentin NH, Young E, Dohlman HG. Systematic analysis of F-box proteins reveals a new branch of the yeast mating pathway. J Biol Chem 2019; 294:14717-14731. [PMID: 31399514 DOI: 10.1074/jbc.ra119.010063] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 08/06/2019] [Indexed: 11/06/2022] Open
Abstract
The mating pathway in yeast Saccharomyces cerevisiae has long been used to reveal new mechanisms of signal transduction. The pathway comprises a pheromone receptor, a heterotrimeric G protein, and intracellular effectors of morphogenesis and transcription. Polarized cell growth, in the direction of a potential mating partner, is accomplished by the G-protein βγ subunits and the small G-protein Cdc42. Transcription induction, needed for cell-cell fusion, is mediated by Gβγ and the mitogen-activated protein kinase (MAPK) scaffold protein Ste5. A potential third pathway is initiated by the G-protein α subunit Gpa1. Gpa1 signaling was shown previously to involve the F-box adaptor protein Dia2 and an endosomal effector protein, the phosphatidylinositol 3-kinase Vps34. Vps34 is also required for proper vacuolar sorting and autophagy. Here, using a panel of reporter assays, we demonstrate that mating pheromone stimulates vacuolar targeting of a cytoplasmic reporter protein and that this process depends on Vps34. Through a systematic analysis of F-box deletion mutants, we show that Dia2 is required to sustain pheromone-induced vacuolar targeting. We also found that other F-box proteins selectively regulate morphogenesis (Ydr306, renamed Pfu1) and transcription (Ucc1). These findings point to the existence of a new and distinct branch of the pheromone-signaling pathway, one that likely leads to vacuolar engulfment of cytoplasmic proteins and recycling of cellular contents in preparation for mating.
Collapse
Affiliation(s)
- Nambirajan Rangarajan
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Claire L Gordy
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Lauren Askew
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Samantha M Bevill
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Timothy C Elston
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Beverly Errede
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Jillian H Hurst
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Joshua B Kelley
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Joshua B Sheetz
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Sara Kimiko Suzuki
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599.,Curriculum in Bioinformatics and Computational Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Natalie H Valentin
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Everett Young
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Henrik G Dohlman
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| |
Collapse
|
152
|
Machado SR, Rodrigues TM. Autophagy and vacuolar biogenesis during the nectary development. PLANTA 2019; 250:519-533. [PMID: 31104130 DOI: 10.1007/s00425-019-03190-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 05/14/2019] [Indexed: 05/26/2023]
Abstract
Different autophagy pathways are a driver of vacuolar biogenesis and are development stage specific during the extrafloral nectary development in Citharexylum myrianthum. Plant autophagy plays an important role in various developmental processes such as seed germination, pollen maturation and leaf senescence. However, studies that address the evidence of autophagy and its role in the development of plant glands are scarce and largely restricted to laticifers. Regarding nectary, studies have repeatedly pointed to signs of degradation associated with the end of the secretory cycle, without exploring autophagy. Likewise, the relationship between autophagy and biogenesis of vacuoles remains an unexplored issue. In this study, using conventional and microwave fixation in association with ultracytochemical methods for transmission electron microscopy, we investigated the occurrence of autophagy and its implication in the differentiation of extrafloral nectary in Citharexylum myrianthum (Verbenaceae) under natural conditions, focusing on the vacuole biogenesis. We described a variety of vacuole types associated with the stage of nectary epidermis development, which differs with respect to origin, function and nature of the products to be stored. Three distinct autophagy pathways were detected: macroautophagy, microautophagy (both restricted to the undifferentiated epidermal cells, at the presecretory stage) and megaautophagy (circumscribed to the differentiated epidermal cells, at the postsecretory stage). Our study clearly demonstrated that the vacuole variety and autophagy processes in the nectary epidermal cells are development specific. This study highlights the role of autophagy in vacuole biogenesis and its implications for the development of nectary and opens new venues for future studies on regulation mechanisms for autophagy in plant secretory structures under normal conditions.
Collapse
Affiliation(s)
- Silvia R Machado
- Department of Botany, Institute of Biosciences of Botucatu (IBB), São Paulo State University (UNESP), Botucatu City, SP, Brazil.
- Center of Electron Microscopy (CME), Institute of Biosciences of Botucatu (IBB), São Paulo State University (UNESP), Botucatu City, SP, Brazil.
| | - Tatiane M Rodrigues
- Department of Botany, Institute of Biosciences of Botucatu (IBB), São Paulo State University (UNESP), Botucatu City, SP, Brazil
| |
Collapse
|
153
|
Ullah S, Zhang M, Yu H, Mustafa S, Shafiq M, Wei Q, Wang W, Jan M, Mao D. Heat exposure affected the reproductive performance of pregnant mice: Enhancement of autophagy and alteration of subcellular structure in the corpus luteum. Reprod Biol 2019; 19:261-269. [PMID: 31285134 DOI: 10.1016/j.repbio.2019.06.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 06/23/2019] [Accepted: 06/28/2019] [Indexed: 01/07/2023]
Abstract
To investigate whether autophagy and subcellular changes are involved in the corpus luteum after heat exposure, a total of 30 early pregnant mice were divided equally into heat stress (HS) and non-HS (NHS) groups (n = 15). Mice in the HS group were exposed to 40.5 ± 0.2 ℃ for 7 consecutive days. Ovaries were collected for immunohistochemistry (IHC), western blot (WB) analysis and transmission electron microscopy (TEM). Serum was collected to determine progesterone by RIA and uteri were collected to count the implantation sites. Results showed that heat exposure increased rectal temperature, decreased body weight and number of implantation sites. WB analysis revealed that ovarian expression of LC3B and Atg7 was up-regulated, while p62 was down-regulated in the HS group. IHC results demonstrated that ovarian staining intensity of LC3B was more intense in the HS group than that of the NHS group. LC3B was mainly localized in the granulosa cells, oocytes and luteal steroidogenic cells of the HS group. TEM results revealed double-layered separated membranes indicative of autophagosomes in the luteal steroidogenic cells of the HS group. Moreover, TEM showed that the mitochondrial cristae became dearth, structure-less, swollen after HS. Additionally, the nucleus expanded and accumulation of lipid droplets increased after HS. Results also showed that heat exposure decreased serum progesterone level and ovarian P450scc expression. These results indicate that HS enhanced autophagy and altered the subcellular structure of luteal steroidogenic cells, which may contribute to interfering with the maintenance of luteal function in early pregnant mice.
Collapse
Affiliation(s)
- Saif Ullah
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, PR China; Faculty of Veterinary and Animal Science, Lasbela University of Agriculture Water and Marine Science, Uthal, Balochistan, 90150, Pakistan
| | - Maoduo Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, PR China
| | - Hao Yu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, PR China
| | - Sheeraz Mustafa
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, PR China
| | - Muhammad Shafiq
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, PR China
| | - Quanwei Wei
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, PR China
| | - Wei Wang
- National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, PR China
| | - Muhammad Jan
- Faculty of Veterinary and Animal Science, Lasbela University of Agriculture Water and Marine Science, Uthal, Balochistan, 90150, Pakistan
| | - Dagan Mao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, PR China.
| |
Collapse
|
154
|
Waskowicz LR, Zhou J, Landau DJ, Brooks ED, Lim A, Yavarow ZA, Kudo T, Zhang H, Wu Y, Grant S, Young SP, Huat BB, Yen PM, Koeberl DD. Bezafibrate induces autophagy and improves hepatic lipid metabolism in glycogen storage disease type Ia. Hum Mol Genet 2019; 28:143-154. [PMID: 30256948 DOI: 10.1093/hmg/ddy343] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Accepted: 09/21/2018] [Indexed: 12/20/2022] Open
Abstract
Glucose-6-phosphatase α (G6Pase) deficiency, also known as von Gierke's Disease or Glycogen storage disease type Ia (GSD Ia), is characterized by decreased ability of the liver to convert glucose-6-phosphate to glucose leading to glycogen accumulation and hepatosteatosis. Long-term complications of GSD Ia include hepatic adenomas and carcinomas, in association with the suppression of autophagy in the liver. The G6pc-/- mouse and canine models for GSD Ia were treated with the pan-peroxisomal proliferator-activated receptor agonist, bezafibrate, to determine the drug's effect on liver metabolism and function. Hepatic glycogen and triglyceride concentrations were measured and western blotting was performed to investigate pathways affected by the treatment. Bezafibrate decreased liver triglyceride and glycogen concentrations and partially reversed the autophagy defect previously demonstrated in GSD Ia models. Changes in medium-chain acyl-CoA dehydrogenase expression and acylcarnintine flux suggested that fatty acid oxidation was increased and fatty acid synthase expression associated with lipogenesis was decreased in G6pc-/- mice treated with bezafibrate. In summary, bezafibrate induced autophagy in the liver while increasing fatty acid oxidation and decreasing lipogenesis in G6pc-/- mice. It represents a potential therapy for glycogen overload and hepatosteatosis associated with GSD Ia, with beneficial effects that have implications for non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Lauren R Waskowicz
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Jin Zhou
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School Singapore, Singapore, Singapore
| | - Dustin J Landau
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Elizabeth D Brooks
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA.,Division of Laboratory Animal Resources, Duke University Medical Center, Durham, NC, USA
| | - Andrea Lim
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School Singapore, Singapore, Singapore
| | - Zollie A Yavarow
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Tsubasa Kudo
- Faculty of Medicine, Tohoku University, Sendai, Japan
| | - Haoyue Zhang
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Yajun Wu
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Stuart Grant
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Sarah P Young
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Bay Boon Huat
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Paul M Yen
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School Singapore, Singapore, Singapore.,Sarah W. Stedman Nutrition and Metabolism Center, Departments of Medicine and Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - Dwight D Koeberl
- Division of Medical Genetics, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA.,Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| |
Collapse
|
155
|
Pellegrini D, Del Grosso A, Angella L, Giordano N, Dilillo M, Tonazzini I, Caleo M, Cecchini M, McDonnell LA. Quantitative Microproteomics Based Characterization of the Central and Peripheral Nervous System of a Mouse Model of Krabbe Disease. Mol Cell Proteomics 2019; 18:1227-1241. [PMID: 30926673 PMCID: PMC6553931 DOI: 10.1074/mcp.ra118.001267] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/15/2019] [Indexed: 11/06/2022] Open
Abstract
Krabbe disease is a rare, childhood lysosomal storage disorder caused by a deficiency of galactosylceramide beta-galactosidase (GALC). The major effect of GALC deficiency is the accumulation of psychosine in the nervous system and widespread degeneration of oligodendrocytes and Schwann cells, causing rapid demyelination. The molecular mechanisms of Krabbe disease are not yet fully elucidated and a definite cure is still missing. Here we report the first in-depth characterization of the proteome of the Twitcher mouse, a spontaneous mouse model of Krabbe disease, to investigate the proteome changes in the Central and Peripheral Nervous System. We applied a TMT-based workflow to compare the proteomes of the corpus callosum, motor cortex and sciatic nerves of littermate homozygous Twitcher and wild-type mice. More than 400 protein groups exhibited differences in expression and included proteins involved in pathways that can be linked to Krabbe disease, such as inflammatory and defense response, lysosomal proteins accumulation, demyelination, reduced nervous system development and cell adhesion. These findings provide new insights on the molecular mechanisms of Krabbe disease, representing a starting point for future functional experiments to study the molecular pathogenesis of Krabbe disease. Data are available via ProteomeXchange with identifier PXD010594.
Collapse
Affiliation(s)
- Davide Pellegrini
- From ‡NEST, Scuola Normale Superiore, Pisa 56127, Italy
- §Fondazione Pisana per la Scienza ONLUS, 56107 San Giuliano Terme, Pisa, Italy
| | - Ambra Del Grosso
- From ‡NEST, Scuola Normale Superiore, Pisa 56127, Italy
- ¶NEST, Istituto Nanoscienze-CNR, Pisa, Italy
| | | | | | - Marialaura Dilillo
- §Fondazione Pisana per la Scienza ONLUS, 56107 San Giuliano Terme, Pisa, Italy
| | | | | | - Marco Cecchini
- From ‡NEST, Scuola Normale Superiore, Pisa 56127, Italy
- ¶NEST, Istituto Nanoscienze-CNR, Pisa, Italy
| | - Liam A McDonnell
- §Fondazione Pisana per la Scienza ONLUS, 56107 San Giuliano Terme, Pisa, Italy;
- **Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
156
|
Amyloid Beta and Phosphorylated Tau-Induced Defective Autophagy and Mitophagy in Alzheimer's Disease. Cells 2019; 8:cells8050488. [PMID: 31121890 PMCID: PMC6562604 DOI: 10.3390/cells8050488] [Citation(s) in RCA: 332] [Impact Index Per Article: 55.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 05/10/2019] [Accepted: 05/21/2019] [Indexed: 12/22/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disease characterized by memory loss and multiple cognitive impairments. Several decades of intense research have revealed that multiple cellular changes are implicated in the development and progression of AD, including mitochondrial damage, synaptic dysfunction, amyloid beta (Aβ) formation and accumulation, hyperphosphorylated tau (P-Tau) formation and accumulation, deregulated microRNAs, synaptic damage, and neuronal loss in patients with AD. Among these, mitochondrial dysfunction and synaptic damage are early events in the disease process. Recent research also revealed that Aβ and P-Tau-induced defective autophagy and mitophagy are prominent events in AD pathogenesis. Age-dependent increased levels of Aβ and P-Tau reduced levels of several autophagy and mitophagy proteins. In addition, abnormal interactions between (1) Aβ and mitochondrial fission protein Drp1; (2) P-Tau and Drp1; and (3) Aβ and PINK1/parkin lead to an inability to clear damaged mitochondria and other cellular debris from neurons. These events occur selectively in affected AD neurons. The purpose of our article is to highlight recent developments of a Aβ and P-Tau-induced defective autophagy and mitophagy in AD. This article also summarizes several aspects of mitochondrial dysfunction, including abnormal mitochondrial dynamics (increased fission and reduced fusion), defective mitochondrial biogenesis, reduced ATP, increased free radicals and lipid peroxidation, and decreased cytochrome c oxidase (COX) activity and calcium dyshomeostasis in AD pathogenesis. Our article also discusses how reduced levels of Drp1, Aβ, and P-Tau can enhance the clearance of damaged mitochondria and other cellular debris by autophagy and mitophagy mechanisms.
Collapse
|
157
|
Anwar T, Liu X, Suntio T, Marjamäki A, Biazik J, Chan EYW, Varjosalo M, Eskelinen EL. ER-Targeted Beclin 1 Supports Autophagosome Biogenesis in the Absence of ULK1 and ULK2 Kinases. Cells 2019; 8:cells8050475. [PMID: 31108943 PMCID: PMC6562811 DOI: 10.3390/cells8050475] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 05/10/2019] [Accepted: 05/15/2019] [Indexed: 12/14/2022] Open
Abstract
Autophagy transports cytoplasmic material and organelles to lysosomes for degradation and recycling. Beclin 1 forms a complex with several other autophagy proteins and functions in the initiation phase of autophagy, but the exact role of Beclin 1 subcellular localization in autophagy initiation is still unclear. In order to elucidate the role of Beclin 1 localization in autophagosome biogenesis, we generated constructs that target Beclin 1 to the endoplasmic reticulum (ER) or mitochondria. Our results confirmed the proper organelle-specific targeting of the engineered Beclin 1 constructs, and the proper formation of autophagy-regulatory Beclin 1 complexes. The ULK kinases are required for autophagy initiation upstream of Beclin 1, and autophagosome biogenesis is severely impaired in ULK1/ULK2 double knockout cells. We tested whether Beclin 1 targeting facilitated its ability to rescue autophagosome formation in ULK1/ULK2 double knockout cells. ER-targeted Beclin 1 was most effective in the rescue experiments, while mitochondria-targeted and non-targeted Beclin 1 also showed an ability to rescue, but with lower activity. However, none of the constructs was able to increase autophagic flux in the knockout cells. We also showed that wild type Beclin 1 was enriched on the ER during autophagy induction, and that ULK1/ULK2 facilitated the ER-enrichment of Beclin 1 under basal conditions. The results suggest that one of the functions of ULK kinases may be to enhance Beclin 1 recruitment to the ER to drive autophagosome formation.
Collapse
Affiliation(s)
- Tahira Anwar
- Molecular and Integrative Biosciences Research Programme, University of Helsinki, 00014 Helsinki, Finland.
| | - Xiaonan Liu
- Institute of Biotechnology & HiLIFE, University of Helsinki, 00014 Helsinki, Finland.
| | - Taina Suntio
- Institute of Biotechnology, University of Helsinki, 00014 Helsinki, Finland.
| | - Annika Marjamäki
- Molecular and Integrative Biosciences Research Programme, University of Helsinki, 00014 Helsinki, Finland.
| | - Joanna Biazik
- Molecular and Integrative Biosciences Research Programme, University of Helsinki, 00014 Helsinki, Finland.
| | - Edmond Y W Chan
- Department of Biomedical and Molecular Sciences and Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON K7L 3N6, Canada.
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK.
| | - Markku Varjosalo
- Institute of Biotechnology & HiLIFE, University of Helsinki, 00014 Helsinki, Finland.
| | - Eeva-Liisa Eskelinen
- Molecular and Integrative Biosciences Research Programme, University of Helsinki, 00014 Helsinki, Finland.
- Institute of Biomedicine, University of Turku, 20520 Turku, Finland.
| |
Collapse
|
158
|
Jatana N, Ascher DB, Pires DEV, Gokhale RS, Thukral L. Human LC3 and GABARAP subfamily members achieve functional specificity via specific structural modulations. Autophagy 2019; 16:239-255. [PMID: 30982432 DOI: 10.1080/15548627.2019.1606636] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Autophagy is a conserved adaptive cellular pathway essential to maintain a variety of physiological functions. Core components of this machinery are the six human Atg8 orthologs that initiate formation of appropriate protein complexes. While these proteins are routinely used as indicators of autophagic flux, it is presently not possible to discern their individual biological functions due to our inability to predict specific binding partners. In our attempts towards determining downstream effector functions, we developed a computational pipeline to define structural determinants of human Atg8 family members that dictate functional diversity. We found a clear evolutionary separation between human LC3 and GABARAP subfamilies and also defined a novel sequence motif responsible for their specificity. By analyzing known protein structures, we observed that functional modules or microclusters reveal a pattern of intramolecular network, including distinct hydrogen bonding of key residues (F52/Y49; a subset of HP2) that may directly modulate their interaction preferences. Multiple molecular dynamics simulations were performed to characterize how these proteins interact with a common protein binding partner, PLEKHM1. Our analysis showed remarkable differences in binding modes via intrinsic protein dynamics, with PLEKHM1-bound GABARAP complexes showing less fluctuations and higher number of contacts. We further mapped 373 genomic variations and demonstrated that distinct cancer-related mutations are likely to lead to significant structural changes. Our findings present a quantitative framework to establish factors underlying exquisite specificity of human Atg8 proteins, and thus facilitate the design of precise modulators.Abbreviations: Atg: autophagy-related; ECs: evolutionary constraints; GABARAP: GABA type A receptor-associated protein; HsAtg8: human Atg8; HP: hydrophobic pocket; KBTBD6: kelch repeat and BTB domain containing 6; LIR: LC3-interacting region; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MD: molecular dynamics; HIV-1 Nef: human immunodeficiency virus type 1 negative regulatory factor; PLEKHM1: pleckstrin homology and RUN domain containing M1; RMSD: root mean square deviation; SQSTM1/p62: sequestosome 1; WDFY3/ALFY: WD repeat and FYVE domain containing 3.
Collapse
Affiliation(s)
- Nidhi Jatana
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - David B Ascher
- Department of Biochemistry and Molecular Biology, Bio21 Institute, University of Melbourne, Melbourne, Victoria, Australia.,Department of Biochemistry, University of Cambridge, Cambridgeshire, UK.,Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Douglas E V Pires
- Department of Biochemistry and Molecular Biology, Bio21 Institute, University of Melbourne, Melbourne, Victoria, Australia.,Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Rajesh S Gokhale
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India.,National Institute of Immunology, New Delhi, India
| | - Lipi Thukral
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR- Institute of Genomics and Integrative Biology, New Delhi, India.,Interdisciplinary Center for Scientific Computing, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
159
|
Mahapatra KK, Panigrahi DP, Praharaj PP, Bhol CS, Patra S, Mishra SR, Behera BP, Bhutia SK. Molecular interplay of autophagy and endocytosis in human health and diseases. Biol Rev Camb Philos Soc 2019; 94:1576-1590. [PMID: 30989802 DOI: 10.1111/brv.12515] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 03/27/2019] [Accepted: 03/29/2019] [Indexed: 12/14/2022]
Abstract
Autophagy, an evolutionarily conserved process for maintaining the physio-metabolic equilibrium of cells, shares many common effector proteins with endocytosis. For example, tethering proteins involved in fusion like Ras-like GTPases (Rabs), soluble N-ethylmaleimide sensitive factor attachment protein receptors (SNAREs), lysosomal-associated membrane protein (LAMP), and endosomal sorting complex required for transport (ESCRT) have a dual role in endocytosis and autophagy, and the trafficking routes of these processes converge at lysosomes. These common effectors indicate an association between budding and fusion of membrane-bound vesicles that may have a substantial role in autophagic lysosome reformation, by sensing cellular stress levels. Therefore, autophagy-endocytosis crosstalk may be significant and implicates a novel endocytic regulatory pathway of autophagy. Moreover, endocytosis has a pivotal role in the intake of signalling molecules, which in turn activates cascades that can result in pathophysiological conditions. This review discusses the basic mechanisms of this crosstalk and its implications in order to identify potential novel therapeutic targets for various human diseases.
Collapse
Affiliation(s)
- Kewal K Mahapatra
- Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India
| | - Debasna P Panigrahi
- Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India
| | - Prakash P Praharaj
- Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India
| | - Chandra S Bhol
- Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India
| | - Srimanta Patra
- Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India
| | - Soumya R Mishra
- Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India
| | - Bishnu P Behera
- Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India
| | - Sujit K Bhutia
- Department of Life Science, National Institute of Technology Rourkela, Sundergarh, Odisha 769008, India
| |
Collapse
|
160
|
Catanese A, Olde Heuvel F, Mulaw M, Demestre M, Higelin J, Barbi G, Freischmidt A, Weishaupt JH, Ludolph AC, Roselli F, Boeckers TM. Retinoic acid worsens ATG10-dependent autophagy impairment in TBK1-mutant hiPSC-derived motoneurons through SQSTM1/p62 accumulation. Autophagy 2019; 15:1719-1737. [PMID: 30939964 DOI: 10.1080/15548627.2019.1589257] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mutations in the TBK1 (TANK binding kinase 1) gene are causally linked to amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). TBK1 phosphorylates the cargo receptors OPTN and SQSTM1 regulating a critical step in macroautophagy/autophagy. Disruption of the autophagic flux leads to accumulation of cytosolic protein aggregates, which are a hallmark of ALS. hiPSC-derived TBK1-mutant motoneurons (MNs) showed reduced TBK1 levels and accumulation of cytosolic SQSTM1-positive aggresomes. By screening a library of nuclear-receptor-agonists for modifiers of the SQSTM1 aggregates, we identified 4-hydroxy(phenyl)retinamide (4HPR) as a potent modifier exerting detrimental effects on mutant-TBK1 motoneurons fitness exacerbating the autophagy overload. We have shown by TEM that TBK1-mutant motoneurons accumulate immature phagophores due a failure in the elongation phase, and 4HPR further worsens the burden of dysfunctional phagophores. 4HPR-increased toxicity was associated with the upregulation of SQSTM1 in a context of strongly reduced ATG10, while rescue of ATG10 levels abolished 4HPR toxicity. Finally, we showed that 4HPR leads to a downregulation of ATG10 and to an accumulation of SQSTM1+ aggresomes also in hiPSC-derived C9orf72-mutant motoneurons. Our data show that cultured human motoneurons harboring mutations in TBK1 gene display typical ALS features, like decreased viability and accumulation of cytosolic SQSTM1-positive aggresomes. The retinoid 4HPR appears a strong negative modifier of the fitness of TBK1 and C9orf72-mutant MNs, through a pathway converging on the mismatch of initiated autophagy and ATG10 levels. Thus, autophagy induction appears not to be a therapeutic strategy for ALS unless the specific underlying pathway alterations are properly addressed. Abbreviations: 4HPR: 4-hydroxy(phenyl)retinamide; AKT: AKT1 serine/threonine kinase 1; ALS: amyotrophic lateral sclerosis; ATG: autophagy related; AVs: autophagic vesicle; C9orf72: chromosome 9 open reading frame 72; CASP3: caspase 3; CHAT: choline O-acetyltransferase; CYCS: cytochrome c, somatic; DIV: day in vitro; FTD: frontotemporal dementia; FUS: FUS RNA binding protein; GFP: green fluorescent protein; hiPSCs: human induced pluripotent stem cells; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MNs: motoneurons; mRFP: monomeric red fluorescent protein; MTOR: mechanistic target of rapamycin kinase; NFE2L2/NRF2: nuclear factor, erythroid 2 like 2; RARA: retinoic acid receptor alpha; SLC18A3/VACHT: solute carrier family 18 (vesicular acetylcholine transporter), member 3; SQSTM1/p62: sequestosome 1; TBK1: TANK binding kinase 1; TEM: transmission electron microscopy.
Collapse
Affiliation(s)
- Alberto Catanese
- Institute of Anatomy and Cell Biology, Ulm University , Ulm , DE , Germany.,International Graduate School, Ulm University , Ulm , DE , Germany
| | | | - Medhanie Mulaw
- Institute of Experimental Tumor Research, Ulm University , Ulm , DE , Germany
| | - Maria Demestre
- Institute of Anatomy and Cell Biology, Ulm University , Ulm , DE , Germany
| | - Julia Higelin
- Institute of Anatomy and Cell Biology, Ulm University , Ulm , DE , Germany.,International Graduate School, Ulm University , Ulm , DE , Germany
| | - Gotthold Barbi
- Institute for Human Genetics, Ulm University , Ulm , DE , Germany
| | | | | | | | - Francesco Roselli
- Institute of Anatomy and Cell Biology, Ulm University , Ulm , DE , Germany.,Deptartment of Neurology, Ulm University , Ulm , DE , Germany
| | - Tobias M Boeckers
- Institute of Anatomy and Cell Biology, Ulm University , Ulm , DE , Germany
| |
Collapse
|
161
|
Autophagy as a Homeostatic Mechanism in Response to Stress Conditions in the Central Nervous System. Mol Neurobiol 2019; 56:6594-6608. [DOI: 10.1007/s12035-019-1546-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 03/12/2019] [Indexed: 12/11/2022]
|
162
|
Guo C, Wang L, Li X, Wang S, Yu X, Xu K, Zhao Y, Luo J, Li X, Jiang B, Shi D. Discovery of Novel Bromophenol-Thiosemicarbazone Hybrids as Potent Selective Inhibitors of Poly(ADP-ribose) Polymerase-1 (PARP-1) for Use in Cancer. J Med Chem 2019; 62:3051-3067. [PMID: 30844273 DOI: 10.1021/acs.jmedchem.8b01946] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Poly(ADP-ribose) polymerase-1 (PARP-1) is a new potential target for anticancer drug discovery. A series of bromophenol-thiosemicarbazone hybrids as PARP-1 inhibitors were designed, synthesized, and evaluated for their antitumor activities. Among them, the most promising compound, 11, showed excellent selective PARP-1 inhibitory activity (IC50 = 29.5 nM) over PARP-2 (IC50 > 1000 nM) and potent anticancer activities toward the SK-OV-3, Bel-7402 and HepG2 cancer cell lines (IC50 = 2.39, 5.45, and 4.60 μM), along with inhibition of tumor growth in an in vivo SK-OV-3 cell xenograft model. Further study demonstrated that compound 11 played an antitumor role through multiple anticancer mechanisms, including the induction of apoptosis and cell cycle arrest, cellular accumulation of DNA double-strand breaks, DNA repair alterations, inhibition of H2O2-triggered PARylation, antiproliferative effects via the production of cytotoxic reactive oxygen species, and autophagy. In addition, compound 11 displayed good pharmacokinetic characteristics and favorable safety. These observations demonstrate that compound 11 may serve as a lead compound for the discovery of new anticancer drugs.
Collapse
Affiliation(s)
- Chuanlong Guo
- Key Laboratory of Experimental Marine Biology , Institute of Oceanology, Chinese Academy of Sciences , Qingdao 266071 , Shandong , China.,Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology , Qingdao 266071 , Shandong , China.,Center for Ocean Mega-Science , Chinese Academy of Sciences , Qingdao 266071 , Shandong , China.,Department of Pharmacy, College of Chemical Engineering , Qingdao University of Science and Technology , Qingdao 266042 , China.,University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Lijun Wang
- Key Laboratory of Experimental Marine Biology , Institute of Oceanology, Chinese Academy of Sciences , Qingdao 266071 , Shandong , China.,Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology , Qingdao 266071 , Shandong , China.,Center for Ocean Mega-Science , Chinese Academy of Sciences , Qingdao 266071 , Shandong , China
| | - Xiuxue Li
- Key Laboratory of Experimental Marine Biology , Institute of Oceanology, Chinese Academy of Sciences , Qingdao 266071 , Shandong , China.,Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology , Qingdao 266071 , Shandong , China.,Center for Ocean Mega-Science , Chinese Academy of Sciences , Qingdao 266071 , Shandong , China
| | - Shuaiyu Wang
- Key Laboratory of Experimental Marine Biology , Institute of Oceanology, Chinese Academy of Sciences , Qingdao 266071 , Shandong , China.,Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology , Qingdao 266071 , Shandong , China.,Center for Ocean Mega-Science , Chinese Academy of Sciences , Qingdao 266071 , Shandong , China
| | - Xuemin Yu
- Department of Otorhinolaryngology , Qilu Hospital of Shandong University , Qingdao 266000 , Shandong , China
| | - Kuo Xu
- Key Laboratory of Experimental Marine Biology , Institute of Oceanology, Chinese Academy of Sciences , Qingdao 266071 , Shandong , China.,Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology , Qingdao 266071 , Shandong , China.,Center for Ocean Mega-Science , Chinese Academy of Sciences , Qingdao 266071 , Shandong , China
| | - Yue Zhao
- Key Laboratory of Experimental Marine Biology , Institute of Oceanology, Chinese Academy of Sciences , Qingdao 266071 , Shandong , China.,Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology , Qingdao 266071 , Shandong , China.,Center for Ocean Mega-Science , Chinese Academy of Sciences , Qingdao 266071 , Shandong , China
| | - Jiao Luo
- Key Laboratory of Experimental Marine Biology , Institute of Oceanology, Chinese Academy of Sciences , Qingdao 266071 , Shandong , China.,Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology , Qingdao 266071 , Shandong , China.,Center for Ocean Mega-Science , Chinese Academy of Sciences , Qingdao 266071 , Shandong , China.,University of Chinese Academy of Sciences , Beijing 100049 , China
| | - Xiangqian Li
- Key Laboratory of Experimental Marine Biology , Institute of Oceanology, Chinese Academy of Sciences , Qingdao 266071 , Shandong , China.,Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology , Qingdao 266071 , Shandong , China.,Center for Ocean Mega-Science , Chinese Academy of Sciences , Qingdao 266071 , Shandong , China
| | - Bo Jiang
- Key Laboratory of Experimental Marine Biology , Institute of Oceanology, Chinese Academy of Sciences , Qingdao 266071 , Shandong , China.,Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology , Qingdao 266071 , Shandong , China.,Center for Ocean Mega-Science , Chinese Academy of Sciences , Qingdao 266071 , Shandong , China
| | - Dayong Shi
- Key Laboratory of Experimental Marine Biology , Institute of Oceanology, Chinese Academy of Sciences , Qingdao 266071 , Shandong , China.,Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology , Qingdao 266071 , Shandong , China.,State Key Laboratory of Microbial Technology , Shandong University , Jinan 250100 , Shandong , China.,Center for Ocean Mega-Science , Chinese Academy of Sciences , Qingdao 266071 , Shandong , China
| |
Collapse
|
163
|
Brier LW, Ge L, Stjepanovic G, Thelen AM, Hurley JH, Schekman R. Regulation of LC3 lipidation by the autophagy-specific class III phosphatidylinositol-3 kinase complex. Mol Biol Cell 2019; 30:1098-1107. [PMID: 30811270 PMCID: PMC6724508 DOI: 10.1091/mbc.e18-11-0743] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Autophagy is a conserved eukaryotic pathway critical for cellular adaptation to changes in nutrition levels and stress. The class III phosphatidylinositol (PI)3-kinase complexes I and II (PI3KC3-C1 and -C2) are essential for autophagosome initiation and maturation, respectively, from highly curved vesicles. We used a cell-free reaction that reproduces a key autophagy initiation step, LC3 lipidation, as a biochemical readout to probe the role of autophagy-related gene (ATG)14, a PI3KC3-C1-specific subunit implicated in targeting the complex to autophagy initiation sites. We reconstituted LC3 lipidation with recombinant PI3KC3-C1, -C2, or various mutant derivatives added to extracts derived from a CRISPR/Cas9-generated ATG14-knockout cell line. Both complexes C1 and C2 require the C-terminal helix of VPS34 for activity on highly curved membranes. However, only complex C1 supports LC3 lipidation through the curvature-targeting amphipathic lipid packing sensor (ALPS) motif of ATG14. Furthermore, the ALPS motif and VPS34 catalytic activity are required for downstream recruitment of WD-repeat domain phosphoinositide-interacting protein (WIPI)2, a protein that binds phosphatidylinositol 3-phosphate and its product phosphatidylinositol 3, 5-bisphosphate, and a WIPI-binding protein, ATG2A, but do not affect membrane association of ATG3 and ATG16L1, enzymes contributing directly to LC3 lipidation. These data reveal the nuanced role of the ATG14 ALPS in membrane curvature sensing, suggesting that the ALPS has additional roles in supporting LC3 lipidation.
Collapse
Affiliation(s)
- Livia W Brier
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94270.,Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94270
| | - Liang Ge
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94270.,Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94270
| | - Goran Stjepanovic
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94270.,California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA 94270.,Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
| | - Ashley M Thelen
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94270
| | - James H Hurley
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94270.,California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA 94270.,Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
| | - Randy Schekman
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94270.,Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94270
| |
Collapse
|
164
|
Kim WJ, Mai A, Weyand NJ, Rendón MA, Van Doorslaer K, So M. Neisseria gonorrhoeae evades autophagic killing by downregulating CD46-cyt1 and remodeling lysosomes. PLoS Pathog 2019; 15:e1007495. [PMID: 30753248 PMCID: PMC6388937 DOI: 10.1371/journal.ppat.1007495] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 02/25/2019] [Accepted: 12/01/2018] [Indexed: 12/20/2022] Open
Abstract
The Gram-negative human pathogen N. gonorrhoeae (Ngo) quickly attaches to epithelial cells, and large numbers of the bacteria remain on the cell surface for prolonged periods. Ngo invades cells but few viable intracellular bacteria are recovered until later stages of infection, leading to the assumption that Ngo is a weak invader. On the cell surface, Ngo quickly recruits CD46-cyt1 to the epithelial cell cortex directly beneath the bacteria and causes its cleavage by metalloproteinases and Presenilin/γSecretease; how these interactions affect the Ngo lifecycle is unknown. Here, we show Ngo induces an autophagic response in the epithelial cell through CD46-cyt1/GOPC, and this response kills early invaders. Throughout infection, the pathogen slowly downregulates CD46-cyt1 and remodeling of lysosomes, another key autophagy component, and these activities ultimately promote intracellular survival. We present a model on the dynamics of Ngo infection and describe how this dual interference with the autophagic pathway allows late invaders to survive within the cell.
Collapse
Affiliation(s)
- Won J. Kim
- BIO5 Institute, University of Arizona, Tucson, AZ, United States of America
- Department of Immunobiology, University of Arizona, Tucson, AZ, United States of America
- * E-mail:
| | - Annette Mai
- BIO5 Institute, University of Arizona, Tucson, AZ, United States of America
| | - Nathan J. Weyand
- Department of Biological Sciences, Ohio University, Athens, OH, United States of America
| | - Maria A. Rendón
- BIO5 Institute, University of Arizona, Tucson, AZ, United States of America
- Department of Immunobiology, University of Arizona, Tucson, AZ, United States of America
| | - Koenraad Van Doorslaer
- BIO5 Institute, University of Arizona, Tucson, AZ, United States of America
- Department of Immunobiology, University of Arizona, Tucson, AZ, United States of America
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, United States of America
| | - Magdalene So
- BIO5 Institute, University of Arizona, Tucson, AZ, United States of America
- Department of Immunobiology, University of Arizona, Tucson, AZ, United States of America
| |
Collapse
|
165
|
Abstract
Many diseases are related to age, among these neurodegeneration is particularly important. Alzheimer's disease Parkinson's and Glaucoma have many common pathogenic events including oxidative damage, Mitochondrial dysfunction, endothelial alterations and changes in the visual field. These are well known in the case of glaucoma, less in the case of neurodegeneration of the brain. Many other molecular aspects are common, such as the role of endoplasmic reticulum autophagy and neuronal apoptosis while others have been neglected due to lack of space such as inflammatory cytokine or miRNA. Moreover, the loss of specific neuronal populations, the induction of similar mechanisms of cell injury and the deposition of protein aggregates in specific anatomical areas are very similar events between these diseases. Intracellular and/or extracellular accumulation of protein aggregates is a key feature of many neurodegenerative disorders. The existence of abnormal protein aggregates has been documented in the RGCs of glaucomatous patients such as the anomalous Tau protein or the β-amyloid accumulations. Intra-cell catabolic processes also appear to be common in both glaucoma and neurodegeneration. They also help us to understand how the basis between these diseases is common and how the visual aspects can be a serious problem for those who are affected.
Collapse
Affiliation(s)
- Sergio Claudio Saccà
- Department of Head/Neck Pathologies, St Martino Hospital, Ophthalmology Unit, Genoa, Italy.
| | - Carlo Alberto Cutolo
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics and Maternal and Child Science, University of Genoa, Policlinico San Martino Hospital, Eye Clinic Genoa, Genoa, Italy
| | - Tommaso Rossi
- Department of Head/Neck Pathologies, St Martino Hospital, Ophthalmology Unit, Genoa, Italy
| |
Collapse
|
166
|
Gassen NC, Rein T. Is There a Role of Autophagy in Depression and Antidepressant Action? Front Psychiatry 2019; 10:337. [PMID: 31156481 PMCID: PMC6529564 DOI: 10.3389/fpsyt.2019.00337] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 04/29/2019] [Indexed: 12/12/2022] Open
Abstract
Autophagy has been recognized as evolutionary conserved intracellular pathway that ensures energy, organelle, and protein homeostasis through lysosomal degradation of damaged macromolecules and organelles. It is activated under various stress situations, e.g., food deprivation or proteotoxic conditions. Autophagy has been linked to several diseases, more recently also including stress-related diseases such as depression. A growing number of publications report on the role of autophagy in neurons, also referred to as "neuronal autophagy" on the one hand, and several studies describe effects of antidepressants-or of compounds that exert antidepressant-like actions-on autophagy on the other hand. This minireview highlights the emerging evidence for the involvement of autophagy in the pathology and treatment of depression and discusses current limitations as well as potential avenues for future research.
Collapse
Affiliation(s)
- Nils C Gassen
- Department of Psychiatry, Bonn Clinical Center, Bonn, Germany.,Max Planck Institute of Psychiatry, Munich, Germany
| | - Theo Rein
- Max Planck Institute of Psychiatry, Munich, Germany
| |
Collapse
|
167
|
Zhao XC, Livingston MJ, Liang XL, Dong Z. Cell Apoptosis and Autophagy in Renal Fibrosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1165:557-584. [PMID: 31399985 DOI: 10.1007/978-981-13-8871-2_28] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Renal fibrosis is the final common pathway of all chronic kidney diseases progressing to end-stage renal diseases. Autophagy, a highly conserved lysosomal degradation pathway, plays important roles in maintaining cellular homeostasis in all major types of kidney cells including renal tubular cells as well as podocytes, mesangial cells and endothelial cells in glomeruli. Autophagy dysfunction is implicated in the pathogenesis of various renal pathologies. Here, we analyze the pathological role and regulation of autophagy in renal fibrosis and related kidney diseases in both glomeruli and tubulointerstitial compartments. Further research is expected to gain significant mechanistic insights and discover pathway-specific and kidney-selective therapies targeting autophagy to prevent renal fibrosis and related kidney diseases.
Collapse
Affiliation(s)
- Xing-Chen Zhao
- Division of Nephrology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Man J Livingston
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University and Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA
| | - Xin-Ling Liang
- Division of Nephrology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.
| | - Zheng Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University and Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA.
| |
Collapse
|
168
|
Weiskirchen R, Tacke F. Relevance of Autophagy in Parenchymal and Non-Parenchymal Liver Cells for Health and Disease. Cells 2019; 8:E16. [PMID: 30609663 PMCID: PMC6357193 DOI: 10.3390/cells8010016] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 12/21/2018] [Accepted: 12/26/2018] [Indexed: 02/06/2023] Open
Abstract
Autophagy is a highly conserved intracellular process for the ordered degradation and recycling of cellular components in lysosomes. In the liver, parenchymal cells (i.e., mainly hepatocytes) utilize autophagy to provide amino acids, glucose, and free fatty acids as sources of energy and biosynthesis functions, but also for recycling and controlling organelles such as mitochondria. Non-parenchymal cells of the liver, including endothelial cells, macrophages (Kupffer cells), and hepatic stellate cells (HSC), also employ autophagy, either for maintaining cellular homeostasis (macrophages, endothelium) or for providing energy for their activation (stellate cells). In hepatocytes, autophagy contributes to essential homeostatic functions (e.g., gluconeogenesis, glycogenolysis, fatty acid oxidation), but is also implicated in diseases. For instance, storage disorders (alpha 1 antitrypsin deficiency, Wilson's disease), metabolic (non-alcoholic steatohepatitis, NASH), and toxic (alcohol) liver diseases may benefit from augmenting autophagy in hepatocytes. In hepatic fibrosis, autophagy has been implicated in the fibrogenic activation of HSC to collagen-producing myofibroblasts. In hepatocellular carcinoma (HCC), autophagy may contribute to tumor surveillance as well as invasiveness, indicating a dual and stage-dependent function in cancer. As many drugs directly or indirectly modulate autophagy, it is intriguing to investigate autophagy-targeting, possibly even cell type-directed strategies for the treatment of hereditary liver diseases, NASH, fibrosis, and HCC.
Collapse
Affiliation(s)
- Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, University Hospital RWTH Aachen, D-52074 Aachen, Germany.
| | - Frank Tacke
- Department of Medicine III, University Hospital RWTH Aachen, D-52074 Aachen, Germany.
| |
Collapse
|
169
|
Autophagy: An Essential Degradation Program for Cellular Homeostasis and Life. Cells 2018; 7:cells7120278. [PMID: 30572663 PMCID: PMC6315530 DOI: 10.3390/cells7120278] [Citation(s) in RCA: 263] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 12/18/2018] [Accepted: 12/18/2018] [Indexed: 12/21/2022] Open
Abstract
Autophagy is a lysosome-dependent cellular degradation program that responds to a variety of environmental and cellular stresses. It is an evolutionarily well-conserved and essential pathway to maintain cellular homeostasis, therefore, dysfunction of autophagy is closely associated with a wide spectrum of human pathophysiological conditions including cancers and neurodegenerative diseases. The discovery and characterization of the kingdom of autophagy proteins have uncovered the molecular basis of the autophagy process. In addition, recent advances on the various post-translational modifications of autophagy proteins have shed light on the multiple layers of autophagy regulatory mechanisms, and provide novel therapeutic targets for the treatment of the diseases.
Collapse
|
170
|
Pupyshev AB, Korolenko TA, Tikhonova MA. Effects and Mechanisms of Rapamycin Action on Experimental Neurodegeneration. NEUROCHEM J+ 2018. [DOI: 10.1134/s1819712418030108] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
171
|
Rowlands J, Heng J, Newsholme P, Carlessi R. Pleiotropic Effects of GLP-1 and Analogs on Cell Signaling, Metabolism, and Function. Front Endocrinol (Lausanne) 2018; 9:672. [PMID: 30532733 PMCID: PMC6266510 DOI: 10.3389/fendo.2018.00672] [Citation(s) in RCA: 192] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 10/29/2018] [Indexed: 12/22/2022] Open
Abstract
The incretin hormone Glucagon-Like Peptide-1 (GLP-1) is best known for its "incretin effect" in restoring glucose homeostasis in diabetics, however, it is now apparent that it has a broader range of physiological effects in the body. Both in vitro and in vivo studies have demonstrated that GLP-1 mimetics alleviate endoplasmic reticulum stress, regulate autophagy, promote metabolic reprogramming, stimulate anti-inflammatory signaling, alter gene expression, and influence neuroprotective pathways. A substantial body of evidence has accumulated with respect to how GLP-1 and its analogs act to restore and maintain normal cellular functions. These findings have prompted several clinical trials which have reported GLP-1 analogs improve cardiac function, restore lung function and reduce mortality in patients with obstructive lung disease, influence blood pressure and lipid storage, and even prevent synaptic loss and neurodegeneration. Mechanistically, GLP-1 elicits its effects via acute elevation in cAMP levels, and subsequent protein kinase(s) activation, pathways well-defined in pancreatic β-cells which stimulate insulin secretion in conjunction with elevated Ca2+ and ATP. More recently, new studies have shed light on additional downstream pathways stimulated by chronic GLP-1 exposure, findings which have direct relevance to our understanding of the potential therapeutic effects of longer lasting analogs recently developed for clinical use. In this review, we provide a comprehensive description of the diverse roles for GLP-1 across multiple tissues, describe downstream pathways stimulated by acute and chronic exposure, and discuss novel pleiotropic applications of GLP-1 mimetics in the treatment of human disease.
Collapse
Affiliation(s)
| | | | - Philip Newsholme
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Perth, WA, Australia
| | - Rodrigo Carlessi
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Perth, WA, Australia
| |
Collapse
|
172
|
Clark SG, Graybeal LL, Bhattacharjee S, Thomas C, Bhattacharya S, Cox DN. Basal autophagy is required for promoting dendritic terminal branching in Drosophila sensory neurons. PLoS One 2018; 13:e0206743. [PMID: 30395636 PMCID: PMC6218061 DOI: 10.1371/journal.pone.0206743] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 10/18/2018] [Indexed: 01/22/2023] Open
Abstract
Dendrites function as the primary sites for synaptic input and integration with impairments in dendritic arborization being associated with dysfunctional neuronal circuitry. Post-mitotic neurons require high levels of basal autophagy to clear cytotoxic materials and autophagic dysfunction under native or cellular stress conditions has been linked to neuronal cell death as well as axo-dendritic degeneration. However, relatively little is known regarding the developmental role of basal autophagy in directing aspects of dendritic arborization or the mechanisms by which the autophagic machinery may be transcriptionally regulated to promote dendritic diversification. We demonstrate that autophagy-related (Atg) genes are positively regulated by the homeodomain transcription factor Cut, and that basal autophagy functions as a downstream effector pathway for Cut-mediated dendritic terminal branching in Drosophila multidendritic (md) sensory neurons. Further, loss of function analyses implicate Atg genes in promoting cell type-specific dendritic arborization and terminal branching, while gain of function studies suggest that excessive autophagy leads to dramatic reductions in dendritic complexity. We demonstrate that the Atg1 initiator kinase interacts with the dual leucine zipper kinase (DLK) pathway by negatively regulating the E3 ubiquitin ligase Highwire and positively regulating the MAPKKK Wallenda. Finally, autophagic induction partially rescues dendritic atrophy defects observed in a model of polyglutamine toxicity. Collectively, these studies implicate transcriptional control of basal autophagy in directing dendritic terminal branching and demonstrate the importance of homeostatic control of autophagic levels for dendritic arbor complexity under native or cellular stress conditions.
Collapse
Affiliation(s)
- Sarah G. Clark
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, United States of America
| | - Lacey L. Graybeal
- School of Systems Biology, Krasnow Institute for Advanced Study, George Mason University, Fairfax, Virginia, United States of America
| | - Shatabdi Bhattacharjee
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, United States of America
| | - Caroline Thomas
- School of Systems Biology, Krasnow Institute for Advanced Study, George Mason University, Fairfax, Virginia, United States of America
| | - Surajit Bhattacharya
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, United States of America
| | - Daniel N. Cox
- Neuroscience Institute, Georgia State University, Atlanta, Georgia, United States of America
- School of Systems Biology, Krasnow Institute for Advanced Study, George Mason University, Fairfax, Virginia, United States of America
- * E-mail:
| |
Collapse
|
173
|
Wei F, Duan Y. Crosstalk between Autophagy and Nanomaterials: Internalization, Activation, Termination. ACTA ACUST UNITED AC 2018; 3:e1800259. [PMID: 32627344 DOI: 10.1002/adbi.201800259] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 10/02/2018] [Indexed: 12/12/2022]
Abstract
Nanomaterials (NMs) are comprehensively applied in biomedicine due to their unique physical and chemical properties. Autophagy, as an evolutionarily conserved cellular quality control process, is closely associated with the effect of NMs on cells. In this review, the recent advances in NM-induced/inhibited autophagy (NM-phagy) are summarized, with an aim to present a comprehensive description of the mechanisms of NM-phagy from the perspective of internalization, activation, and termination, thereby bridging autophagy and nanomaterials. Several possible mechanisms are extensively reviewed including the endocytosis pathway of NMs and the related cross components (clathrin and adaptor protein 2 (AP-2), adenosine diphosphate (ADP)-ribosylation factor 6 (Arf6), Rab, UV radiation resistance associated gene (UVRAG)), three main stress mechanisms (oxidative stress, damaged organelles stress, and toxicity stress), and several signal pathway-related molecules. The mechanistic insight is beneficial to understand the autophagic response to NMs or NMs' regulation of autophagy. The challenges currently encountered and research trend in the field of NM-phagy are also highlighted. It is hoped that the NM-phagy discussion in this review with the focus on the mechanistic aspects may serve as a guideline for future research in this field.
Collapse
Affiliation(s)
- Fujing Wei
- Research Center of Analytical Instrumentation, Key Laboratory of Bio-resource and Eco-enviroment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, P. R. China
| | - Yixiang Duan
- Research Center of Analytical Instrumentation, Key Laboratory of Bio-resource and Eco-enviroment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, P. R. China
| |
Collapse
|
174
|
Wu R, Murali R, Kabe Y, French SW, Chiang YM, Liu S, Sher L, Wang CC, Louie S, Tsukamoto H. Baicalein Targets GTPase-Mediated Autophagy to Eliminate Liver Tumor-Initiating Stem Cell-Like Cells Resistant to mTORC1 Inhibition. Hepatology 2018; 68:1726-1740. [PMID: 29729190 PMCID: PMC6204108 DOI: 10.1002/hep.30071] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 04/13/2018] [Accepted: 04/30/2018] [Indexed: 12/30/2022]
Abstract
Drug resistance is a major problem in the treatment of liver cancer. Mammalian Target of Rapamycin 1 (mTORC1) inhibitors have been tested for the treatment of liver cancer based on hyperactive mTOR in this malignancy. However, their clinical trials showed poor outcome, most likely due to their ability to upregulate CD133 and promote chemoresistance. The CD133+ tumor-initiating stem cell-like cells (TICs) isolated from mouse and human liver tumors are chemoresistant, and identification of an approach to abrogate this resistance is desired. In search of a compound that rescinds resistance of TICs to mTORC1 inhibition and improves chemotherapy, we identified baicalein (BC), which selectively chemosensitizes TICs and the human hepatocellular carcinoma (HCC) cell line Huh7 cells but not mouse and human primary hepatocytes. Nanobead pull-down and mass-spectrometric analysis, biochemical binding assay, and three-dimensional computational modeling studies reveal BC's ability to competitively inhibit guanosine triphosphate binding of SAR1B guanosine triphosphatase, which is essential for autophagy. Indeed, BC suppresses autophagy induced by an mTORC1 inhibitor and synergizes cell death caused by mTORC1 inhibition in TIC and Huh7 spheroid formation and in the patient-derived xenograft model of HCC. The BC-induced chemosensitization is rescued by SAR1B expression and phenocopied by SAR1B knockdown in cancer cells treated with a mTORC1 inhibitor. Conclusion: These results identify SAR1B as a target in liver TICs and HCC cells resistant to mTORC1 inhibition.
Collapse
Affiliation(s)
- Raymond Wu
- Southern California Research Center for ALPD and Cirrhosis and Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Ramachandran Murali
- Department of Biomedical Sciences, Cedars Sinai Medical Center, Los Angeles, California, USA
| | - Yasuaki Kabe
- Department of Biochemistry, Keio University of School of Medicine, Tokyo, Japan
| | | | - Yi-Ming Chiang
- School of Pharmacy, University of Southern California, Los Angeles, California, USA
| | - Siyu Liu
- School of Pharmacy, University of Southern California, Los Angeles, California, USA
| | - Linda Sher
- Department of Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Clay C. Wang
- School of Pharmacy, University of Southern California, Los Angeles, California, USA
| | - Stan Louie
- School of Pharmacy, University of Southern California, Los Angeles, California, USA
| | - Hidekazu Tsukamoto
- Southern California Research Center for ALPD and Cirrhosis and Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
- Department of Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, USA
| |
Collapse
|
175
|
Corona AK, Jackson WT. Finding the Middle Ground for Autophagic Fusion Requirements. Trends Cell Biol 2018; 28:869-881. [PMID: 30115558 PMCID: PMC6197918 DOI: 10.1016/j.tcb.2018.07.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 06/29/2018] [Accepted: 07/06/2018] [Indexed: 12/26/2022]
Abstract
Autophagosome/amphisome-lysosome fusion is a highly regulated process at the protein, lipid, and biochemical level. Each primary component of fusion, such as the core SNAREs, HOPS complex, or physical positioning by microtubule-associated dynein motors, are regulated at multiple points to ensure optimum conditions for autophagic flux to proceed. With the complexity of the membrane fusion system, it is not difficult to imagine how autophagic flux defect-related disorders, such as Huntington's disease, non-familial Alzheimer's disease, and Vici syndrome develop. Each membrane fusion step is regulated at the protein, lipid, and ion level. This review aims to discuss the recent developments toward understanding the regulation of autophagosome, amphisome, and lysosome fusion requirements for successful autophagic flux.
Collapse
Affiliation(s)
- Abigail K Corona
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Avenue, Baltimore, MD 21201, USA
| | - William T Jackson
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Avenue, Baltimore, MD 21201, USA.
| |
Collapse
|
176
|
Li L, Liu X, Zhou L, Wang W, Liu Z, Cheng Y, Li J, Wei H. Autophagy Plays a Critical Role in Insulin Resistance- Mediated Chemoresistance in Hepatocellular Carcinoma Cells by Regulating the ER Stress. J Cancer 2018; 9:4314-4324. [PMID: 30519335 PMCID: PMC6277662 DOI: 10.7150/jca.27943] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 09/14/2018] [Indexed: 02/07/2023] Open
Abstract
The high mortality of hepatocellular carcinoma (HCC) patients is associated with several independent risk factors including type 2 diabetes mellitus (T2DM) and insulin resistance (IR), which could be caused by various pathological processes such as tumorigenesis and inflammation in the liver. In previous report, we declared that IR contributes to multidrug resistance in HCC by activation of endoplasmic reticulum (ER) stress. Here, our study revealed that the enhanced autophagy induced by IR significantly prompts the chemotherapeutic drug resistance in hepatoma cells, which was validated by stimulation and inhibition of the autophagy respectively. A potential reason is that autophagy acts as a regulator of ER stress in the IR-mediated chemoresistance in HCC. In conclusion, autophagy facilitates the HCC survival in chemotherapeutic drug treatment by maintaining the homeostasis in the ER indicating the regulatory role of autophagy in ER stress contributes to IR-mediated chemoresistance in hepatocellular carcinoma cells. Collectively, these data implied inhibition of autophagy is a potential treatment of inherent IR-mediated chemoresistance in HCC.
Collapse
Affiliation(s)
- Linjing Li
- Department of Clinical Laboratory Center, The Second Hospital of Lanzhou University, Lanzhou 730000, P.R. China.,Dana-Farber cancer institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02215-5450, USA
| | - Xinyue Liu
- Department of Clinical Laboratory Center, The Second Hospital of Lanzhou University, Lanzhou 730000, P.R. China
| | - Liye Zhou
- Dana-Farber cancer institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA, 02215-5450, USA
| | - Wei Wang
- Marlene and Stewart Greenbaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Zhuan Liu
- Department of Clinical Laboratory Center, The Second Hospital of Lanzhou University, Lanzhou 730000, P.R. China
| | - Yan Cheng
- Northwest University for Nationalities, Lanzhou 730000, P.R. China
| | - Jing Li
- Department of Clinical Laboratory Center, The Second Hospital of Lanzhou University, Lanzhou 730000, P.R. China
| | - Hulai Wei
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, P.R. China
| |
Collapse
|
177
|
Ma K, Wu H, Li P, Li B. LC3-II may mediate ATR-induced mitophagy in dopaminergic neurons through SQSTM1/p62 pathway. Acta Biochim Biophys Sin (Shanghai) 2018; 50:1047-1061. [PMID: 30084861 DOI: 10.1093/abbs/gmy091] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 07/05/2018] [Indexed: 11/12/2022] Open
Abstract
Atrazine (2-chloro-4-ethylamino-6-isopropylamine-1,3,5-triazine; ATR) has been demonstrated to regulate autophagy- and apoptosis-related proteins in doparminergic neuronal damage. In our study, we investigated the role of LC3-II in ATR-induced degeneration of dopaminergic neurons. In vivo dopaminergic neuron degeneration model was set up with ATR treatment and confirmed by the behavioral responses and pathological analysis. Dopaminergic neuron cells were transfected with LC3-II siRNA and treated with ATR to observe cell survival and reactive oxygen species release. The process of mitochondrial autophagy and the neurotoxic effects of mitochondrial autophagy were detected by immunofluorescence assay, immunohistochemical analysis, real-time PCR, and western blot analysis. Results showed that after ATR treatment, the grip strength of Wistar rats was significantly decreased, and behavioral signs of anxiety were clearly observed. The mRNA and protein levels of tyrosine hydroxylase, LC3-II, PINK1, and Parkin were significantly decreased in ATR-induced rat dopaminergic neurons and PC-12 cells, while the mRNA expression and protein levels of SQSTM1/p62 and Parl were increased. Exposure to ATR also led to accumulation of autophagic lysosomes and autophagic bodies along with significantly decreased levels of dopaminergic neurons and alterations in mitochondrial homeostasis, which was reversed by LC3-II siRNA. Our results suggest that ATR affects the mitochondria-mediated dopaminergic neuronal death, which may be mediated by LC3-II and other autophagy markers in vivo and in vitro through SQSTM1/p62 signaling pathway.
Collapse
Affiliation(s)
- Kun Ma
- Department of Hygienic Toxicology, Public Health College, Harbin Medical University, Harbin, China
| | - Haoyu Wu
- Department of Hygienic Toxicology, Public Health College, Harbin Medical University, Harbin, China
| | - Peng Li
- Department of Hygienic Toxicology, Public Health College, Harbin Medical University, Harbin, China
| | - Baixiang Li
- Department of Hygienic Toxicology, Public Health College, Harbin Medical University, Harbin, China
| |
Collapse
|
178
|
Wei Y, Liu M, Li X, Liu J, Li H. Origin of the Autophagosome Membrane in Mammals. BIOMED RESEARCH INTERNATIONAL 2018; 2018:1012789. [PMID: 30345294 PMCID: PMC6174804 DOI: 10.1155/2018/1012789] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 08/18/2018] [Accepted: 09/03/2018] [Indexed: 12/20/2022]
Abstract
Autophagy begins with the nucleation of phagophores, which then expand to give rise to the double-membrane autophagosomes. Autophagosomes ultimately fuse with lysosomes, where the cytosolic cargoes are degraded. Accumulation of autophagosomes is a hallmark of autophagy and neurodegenerative disorders including Alzheimer's and Huntington's disease. In recent years, the sources of autophagosome membrane have attracted a great deal of interests, even so, the membrane donors for autophagosomes are still under debate. In this review, we describe the probable sources of autophagosome membrane.
Collapse
Affiliation(s)
- Yun Wei
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Meixia Liu
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Xianxiao Li
- Department of Oncology, Air Force General Hospital, Beijing 100142, China
| | - Jiangang Liu
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Hao Li
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing 100091, China
| |
Collapse
|
179
|
Paquet C, Nicoll JAR, Love S, Mouton‐Liger F, Holmes C, Hugon J, Boche D. Downregulated apoptosis and autophagy after anti-Aβ immunotherapy in Alzheimer's disease. Brain Pathol 2018; 28:603-610. [PMID: 29027727 PMCID: PMC8028546 DOI: 10.1111/bpa.12567] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 10/08/2017] [Accepted: 10/09/2017] [Indexed: 01/13/2023] Open
Abstract
Aβ immunization of Alzheimer's disease (AD) patients in the AN1792 (Elan Pharmaceuticals) trial caused Aβ removal and a decreased density of neurons in the cerebral cortex. As preservation of neurons may be a critical determinant of outcome after Aβ immunization, we have assessed the impact of previous Aβ immunization on the expression of a range of apoptotic proteins in post-mortem human brain tissue. Cortex from 13 AD patients immunized with AN1792 (iAD) and from 27 nonimmunized AD (cAD) cases was immunolabeled for proapoptotic proteins implicated in AD pathophysiology: phosphorylated c-Jun N-terminal kinase (pJNK), activated caspase3 (a-casp3), phosphorylated GSK3β on tyrosine 216 (GSK3βtyr216 ), p53 and Cdk5/p35. Expression of these proteins was analyzed in relation to immunization status and other clinical data. The antigen load of all of these proapoptotic proteins was significantly lower in iAD than cAD (P < 0.0001). In cAD, significant correlations (P < 0.001) were observed between: Cdk5/p35 and GSK3βtyr216 ; a-casp3 and Aβ42 ; p53 and age at death. In iAD, significant correlations were found between GSK3βtyr216 and a-casp3; both spongiosis and neuritic curvature ratio and Aβ42 ; and Cdk5/p35 and Aβ-antibody level. Although neuronal loss was increased by immunization with AN1792, our present findings suggest downregulation of apoptosis in residual neurons and other cells.
Collapse
Affiliation(s)
- Claire Paquet
- UMRS, INSERM, U942, F‐75010ParisFrance
- University of Paris Diderot, Sorbonne Paris CitéParisFrance
- Centre de Neurologie Cognitive/Centre Memoire de Ressources et de Recherches Paris Nord Ile de France AP‐HP, Hôpital Lariboisière, F‐75010ParisFrance
| | - James AR Nicoll
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of MedicineUniversity of SouthamptonSouthamptonUnited Kingdom
- Department of Cellular PathologyUniversity Hospital Southampton NHS Foundation TrustSouthamptonUnited Kingdom
| | - Seth Love
- Department of Neuropathology, Institute of Clinical Neurosciences, School of Clinical SciencesUniversity of BristolBristolUnited Kingdom
| | - François Mouton‐Liger
- University of Paris Diderot, Sorbonne Paris CitéParisFrance
- Inserm, U1127, Institut du Cerveau et de la Moelle épinière, ICM, F‐75013ParisFrance
| | - Clive Holmes
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of MedicineUniversity of SouthamptonSouthamptonUnited Kingdom
- Memory Assessments and Research Centre, Moorgreen Hospital, Southern Health Foundation TrustSouthampton United Kingdom
| | - Jacques Hugon
- UMRS, INSERM, U942, F‐75010ParisFrance
- University of Paris Diderot, Sorbonne Paris CitéParisFrance
- Centre de Neurologie Cognitive/Centre Memoire de Ressources et de Recherches Paris Nord Ile de France AP‐HP, Hôpital Lariboisière, F‐75010ParisFrance
| | - Delphine Boche
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of MedicineUniversity of SouthamptonSouthamptonUnited Kingdom
| |
Collapse
|
180
|
Robke L, Rodrigues T, Schröder P, Foley DJ, Bernardes GJ, Laraia L, Waldmann H. Discovery of 2,4-dimethoxypyridines as novel autophagy inhibitors. Tetrahedron 2018. [DOI: 10.1016/j.tet.2018.07.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
181
|
Gemcitabine resistance mediated by ribonucleotide reductase M2 in lung squamous cell carcinoma is reversed by GW8510 through autophagy induction. Clin Sci (Lond) 2018; 132:1417-1433. [PMID: 29853661 DOI: 10.1042/cs20180010] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Revised: 05/13/2018] [Accepted: 05/25/2018] [Indexed: 12/28/2022]
Abstract
Although chemotherapeutic regimen containing gemcitabine is the first-line therapy for advanced lung squamous cell carcinoma (LSCC), gemcitabine resistance remains an important clinical problem. Some studies suggest that overexpressions of ribonucleotide reductase (RNR) subunit M2 (RRM2) may be involved in gemcitabine resistance. We used a novel RRM2 inhibitor, GW8510, as a gemcitabine sensitization agent to investigate the therapeutic utility in reversing gemcitabine resistance in LSCC. Results showed that the expressions of RRM2 were increased in gemcitabine intrinsic resistant LSCC cells upon gemcitabine treatment. GW8510 not only suppressed LSCC cell survival, but also sensitized gemcitabine-resistant cells to gemcitabine through autophagy induction mediated by RRM2 down-regulation along with decrease in dNTP levels. The combination of GW8510 and gemcitabine produced a synergistic effect on killing LSCC cells. The synergism of the two agents was impeded by addition of autophagy inhibitors chloroquine (CQ) or bafilomycin A1 (Baf A1), or knockdown of the autophagy gene, Bcl-2-interacting protein 1 (BECN1). Moreover, GW8510-caused LSCC cell sensitization to gemcitabine through autophagy induction was parallel with impairment of DNA double-strand break (DSB) repair and marked increase in cell apoptosis, revealing a cross-talk between autophagy and DNA damage repair, and an interplay between autophagy and apoptosis. Finally, gemcitabine sensitization mediated by autophagy induction through GW8510-caused RRM2 down-regulation was demonstrated in vivo in gemcitabine-resistant LSCC tumor xenograft, further indicating that the sensitization is dependent on autophagy activation. In conclusion, GW8510 can reverse gemcitabine resistance in LSCC cells through RRM2 downregulation-mediated autophagy induction, and GW850 may be a promising therapeutic agent against LSCC as it combined with gemcitabine.
Collapse
|
182
|
Wang Y, Song M, Song F. Neuronal autophagy and axon degeneration. Cell Mol Life Sci 2018; 75:2389-2406. [PMID: 29675785 PMCID: PMC11105516 DOI: 10.1007/s00018-018-2812-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 03/13/2018] [Accepted: 04/03/2018] [Indexed: 02/08/2023]
Abstract
Axon degeneration is a pathophysiological process of axonal dying and breakdown, which is characterized by several morphological features including the accumulation of axoplasmic organelles, disassembly of microtubules, and fragmentation of the axonal cytoskeleton. Autophagy, a highly conserved lysosomal-degradation machinery responsible for the control of cellular protein quality, is widely believed to be essential for the maintenance of axonal homeostasis in neurons. In recent years, more and more evidence suggests that dysfunctional autophagy is associated with axonal degeneration in many neurodegenerative diseases. Here, we review the core machinery of autophagy in neuronal cells, and provide several major steps that interfere with autophagy flux in neurodegenerative conditions. Furthermore, this review highlights the potential role of neuronal autophagy in axon degeneration, and presents some possible molecular mechanisms by which dysfunctional autophagy leads to axon degeneration in pathological conditions.
Collapse
Affiliation(s)
- Yu Wang
- Department of Toxicology, School of Public Health, Shandong University, 44 Wenhuaxi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Mingxue Song
- School of Public Health, Fujian Medical University, 1 Xueyuan Road, Fuzhou, 350108, Fujian, People's Republic of China
| | - Fuyong Song
- Department of Toxicology, School of Public Health, Shandong University, 44 Wenhuaxi Road, Jinan, 250012, Shandong, People's Republic of China.
| |
Collapse
|
183
|
MDA-9/Syntenin regulates protective autophagy in anoikis-resistant glioma stem cells. Proc Natl Acad Sci U S A 2018; 115:5768-5773. [PMID: 29760085 DOI: 10.1073/pnas.1721650115] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Glioma stem cells (GSCs) comprise a small subpopulation of glioblastoma multiforme cells that contribute to therapy resistance, poor prognosis, and tumor recurrence. Protective autophagy promotes resistance of GSCs to anoikis, a form of programmed cell death occurring when anchorage-dependent cells detach from the extracellular matrix. In nonadherent conditions, GSCs display protective autophagy and anoikis-resistance, which correlates with expression of melanoma differentiation associated gene-9/Syntenin (MDA-9) (syndecan binding protein; SDCBP). When MDA-9 is suppressed, GSCs undergo autophagic death supporting the hypothesis that MDA-9 regulates protective autophagy in GSCs under anoikis conditions. MDA-9 maintains protective autophagy through phosphorylation of BCL2 and by suppressing high levels of autophagy through EGFR signaling. MDA-9 promotes these changes by modifying FAK and PKC signaling. Gain-of-function and loss-of-function genetic approaches demonstrate that MDA-9 regulates pEGFR and pBCL2 expression through FAK and pPKC. EGFR signaling inhibits autophagy markers (ATG5, Lamp1, LC3B), helping to maintain protective autophagy, and along with pBCL2 maintain survival of GSCs. In the absence of MDA-9, this protective mechanism is deregulated; EGFR no longer maintains protective autophagy, leading to highly elevated and sustained levels of autophagy and consequently decreased cell survival. In addition, pBCL2 is down-regulated in the absence of MDA-9, leading to cell death in GSCs under conditions of anoikis. Our studies confirm a functional link between MDA-9 expression and protective autophagy in GSCs and show that inhibition of MDA-9 reverses protective autophagy and induces anoikis and cell death in GSCs.
Collapse
|
184
|
Inhibition of stress-inducible HSP70 impairs mitochondrial proteostasis and function. Oncotarget 2018; 8:45656-45669. [PMID: 28484090 PMCID: PMC5542216 DOI: 10.18632/oncotarget.17321] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 04/03/2017] [Indexed: 12/20/2022] Open
Abstract
Protein quality control is an important component of survival for all cells. The use of proteasome inhibitors for cancer therapy derives from the fact that tumor cells generally exhibit greater levels of proteotoxic stress than do normal cells, and thus cancer cells tend to be more sensitive to proteasome inhibition. However, this approach has been limited in some cases by toxicity to normal cells. Recently, the concept of inhibiting proteostasis in organelles for cancer therapy has been advanced, in part because it is predicted to have reduced toxicity for normal cells. Here we demonstrate that a fraction of the major stress-induced chaperone HSP70 (also called HSPA1A or HSP72, but hereafter HSP70) is abundantly present in mitochondria of tumor cells, but is expressed at quite low or undetectable levels in mitochondria of most normal tissues and non-tumor cell lines. We show that treatment of tumor cells with HSP70 inhibitors causes a marked change in mitochondrial protein quality control, loss of mitochondrial membrane potential, reduced oxygen consumption rate, and loss of ATP production. We identify several nuclear-encoded mitochondrial proteins, including polyadenylate binding protein-1 (PABPC1), which exhibit decreased abundance in mitochondria following treatment with HSP70 inhibitors. We also show that targeting HSP70 function leads to reduced levels of several mitochondrial-encoded RNA species that encode components of the electron transport chain. Our data indicate that small molecule inhibitors of HSP70 represent a new class of organelle proteostasis inhibitors that impair mitochondrial function in cancer cells, and therefore constitute novel therapeutics.
Collapse
|
185
|
Wang P, Shao BZ, Deng Z, Chen S, Yue Z, Miao CY. Autophagy in ischemic stroke. Prog Neurobiol 2018; 163-164:98-117. [DOI: 10.1016/j.pneurobio.2018.01.001] [Citation(s) in RCA: 310] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Revised: 12/04/2017] [Accepted: 01/10/2018] [Indexed: 02/07/2023]
|
186
|
Mrakovcic M, Fröhlich LF. p53-Mediated Molecular Control of Autophagy in Tumor Cells. Biomolecules 2018; 8:E14. [PMID: 29561758 PMCID: PMC6022997 DOI: 10.3390/biom8020014] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 03/16/2018] [Accepted: 03/19/2018] [Indexed: 12/19/2022] Open
Abstract
Autophagy is an indispensable mechanism of the eukaryotic cell, facilitating the removal and renewal of cellular components and thereby balancing the cell's energy consumption and homeostasis. Deregulation of autophagy is now regarded as one of the characteristic key features contributing to the development of tumors. In recent years, the suppression of autophagy in combination with chemotherapeutic treatment has been approached as a novel therapy in cancer treatment. However, depending on the type of cancer and context, interference with the autophagic machinery can either promote or disrupt tumorigenesis. Therefore, disclosure of the major signaling pathways that regulate autophagy and control tumorigenesis is crucial. To date, several tumor suppressor proteins and oncogenes have emerged as eminent regulators of autophagy whose depletion or mutation favor tumor formation. The mammalian cell "janitor" p53 belongs to one of these tumor suppressors that are most commonly mutated in human tumors. Experimental evidence over the last decade convincingly reports that p53 can act as either an activator or an inhibitor of autophagy depending on its subcellular localization and its mode of action. This finding gains particular significance as p53 deficiency or mutant variants of p53 that accumulate in the cytoplasm of tumor cells enable activation of autophagy. Accordingly, we recently identified p53 as a molecular hub that regulates autophagy and apoptosis in histone deacetylase inhibitor-treated uterine sarcoma cells. In light of this novel experimental evidence, in this review, we focus on p53 signaling as a mediator of the autophagic pathway in tumor cells.
Collapse
Affiliation(s)
- Maria Mrakovcic
- AG VABOS, Department of Cranio-Maxillofacial Surgery, University of Münster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany.
| | - Leopold F Fröhlich
- AG VABOS, Department of Cranio-Maxillofacial Surgery, University of Münster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany.
| |
Collapse
|
187
|
Rahman MA, Rhim H. Therapeutic implication of autophagy in neurodegenerative diseases. BMB Rep 2018; 50:345-354. [PMID: 28454606 PMCID: PMC5584741 DOI: 10.5483/bmbrep.2017.50.7.069] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Indexed: 12/14/2022] Open
Abstract
Autophagy, a catabolic process necessary for the maintenance of intracellular homeostasis, has recently been the focus of numerous human diseases and conditions, such as aging, cancer, development, immunity, longevity, and neurodegeneration. However, the continued presence of autophagy is essential for cell survival and dysfunctional autophagy is thought to speed up the progression of neurodegeneration. The actual molecular mechanism behind the progression of dysfunctional autophagy is not yet fully understood. Emerging evidence suggests that basal autophagy is necessary for the removal of misfolded, aggregated proteins and damaged cellular organelles through lysosomal mediated degradation. Physiologically, neurodegenerative disorders are related to the accumulation of amyloid β peptide and α-synuclein protein aggregation, as seen in patients with Alzheimer’s disease and Parkinson’s disease, respectively. Even though autophagy could impact several facets of human biology and disease, it generally functions as a clearance for toxic proteins in the brain, which contributes novel insight into the pathophysiological understanding of neurodegenerative disorders. In particular, several studies demonstrate that natural compounds or small molecule autophagy enhancer stimuli are essential in the clearance of amyloid β and α-synuclein deposits. Therefore, this review briefly deliberates on the recent implications of autophagy in neurodegenerative disorder control, and emphasizes the opportunities and potential therapeutic application of applied autophagy.
Collapse
Affiliation(s)
- Md Ataur Rahman
- Center for Neuroscience, Korea Institute of Science and Technology, Seoul 02792, Korea
| | - Hyewhon Rhim
- Center for Neuroscience, Korea Institute of Science and Technology, Seoul 02792; Department of Neuroscience, Korea University of Science and Technology, Daejeon 34113, Korea
| |
Collapse
|
188
|
Castets P, Frank S, Sinnreich M, Rüegg MA. "Get the Balance Right": Pathological Significance of Autophagy Perturbation in Neuromuscular Disorders. J Neuromuscul Dis 2018; 3:127-155. [PMID: 27854220 PMCID: PMC5271579 DOI: 10.3233/jnd-160153] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Recent research has revealed that autophagy, a major catabolic process in cells, is dysregulated in several neuromuscular diseases and contributes to the muscle wasting caused by non-muscle disorders (e.g. cancer cachexia) or during aging (i.e. sarcopenia). From there, the idea arose to interfere with autophagy or manipulate its regulatory signalling to help restore muscle homeostasis and attenuate disease progression. The major difficulty for the development of therapeutic strategies is to restore a balanced autophagic flux, due to the dynamic nature of autophagy. Thus, it is essential to better understand the mechanisms and identify the signalling pathways at play in the control of autophagy in skeletal muscle. A comprehensive analysis of the autophagic flux and of the causes of its dysregulation is required to assess the pathogenic role of autophagy in diseased muscle. Furthermore, it is essential that experiments distinguish between primary dysregulation of autophagy (prior to disease onset) and impairments as a consequence of the pathology. Of note, in most muscle disorders, autophagy perturbation is not caused by genetic modification of an autophagy-related protein, but rather through indirect alteration of regulatory signalling or lysosomal function. In this review, we will present the mechanisms involved in autophagy, and those ensuring its tight regulation in skeletal muscle. We will then discuss as to how autophagy dysregulation contributes to the pathogenesis of neuromuscular disorders and possible ways to interfere with this process to limit disease progression.
Collapse
Affiliation(s)
| | - Stephan Frank
- Institute of Pathology, Division of Neuropathology Basel University Hospital, Basel, Switzerland
| | - Michael Sinnreich
- Neuromuscular Research Center, Departments of Neurology and Biomedicine, Pharmazentrum, Basel, Switzerland
| | | |
Collapse
|
189
|
Gosavi P, Houghton FJ, McMillan PJ, Hanssen E, Gleeson PA. The Golgi ribbon in mammalian cells negatively regulates autophagy by modulating mTOR activity. J Cell Sci 2018; 131:jcs.211987. [PMID: 29361552 DOI: 10.1242/jcs.211987] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 12/20/2017] [Indexed: 12/25/2022] Open
Abstract
In vertebrates, individual Golgi stacks are joined into a compact ribbon structure; however, the relevance of a ribbon structure has been elusive. Here, we exploit the finding that the membrane tether of the trans-Golgi network, GCC88 (encoded by GCC1), regulates the balance between Golgi mini-stacks and the Golgi ribbon. Loss of Golgi ribbons in stable cells overexpressing GCC88 resulted in compromised mechanistic target of rapamycin (mTOR) signaling and a dramatic increase in LC3-II-positive autophagosomes, whereas RNAi-mediated depletion of GCC88 restored the Golgi ribbon and reduced autophagy. mTOR was absent from dispersed Golgi mini-stacks whereas recruitment of mTOR to lysosomes was unaffected. We show that the Golgi ribbon is a site for localization and activation of mTOR, a process dependent on the ribbon structure. We demonstrate a strict temporal sequence of fragmentation of Golgi ribbon, loss of Golgi mTOR and subsequent increased autophagy. Golgi ribbon fragmentation has been reported in various neurodegenerative diseases and we demonstrate the potential relevance of our findings in neuronal cells using a model of neurodegeneration. Overall, this study highlights a role for the Golgi ribbon in pathways central to cellular homeostasis.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Prajakta Gosavi
- The Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Fiona J Houghton
- The Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Paul J McMillan
- The Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria 3010, Australia.,Biological Optical Microscopy Platform, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Eric Hanssen
- The Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria 3010, Australia.,Advanced Microscopy Facility, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Paul A Gleeson
- The Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria 3010, Australia
| |
Collapse
|
190
|
Chen Y, Yu L. Development of Research into Autophagic Lysosome Reformation. Mol Cells 2018; 41:45-49. [PMID: 29370688 PMCID: PMC5792712 DOI: 10.14348/molcells.2018.2265] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 11/30/2017] [Indexed: 12/17/2022] Open
Abstract
Autophagy is a lysosome-dependent degradation process that is essential for maintaining cellular homeostasis. In recent years, more studies have focused on the late stages of autophagy. Our group discovered and studied the terminal step of autophagy, namely autophagic lysosome reformation (ALR). ALR is the process that regenerates functional lysosomes from autolysosomes, thus maintaining lysosome homeostasis. ALR involves clathrin-mediated membrane budding from autolysosomes, elongation of membrane tubules along microtubules with the pulling force provided by the motor protein KIF5B, proto-lysosome scission by dynamin 2, and finally maturation of proto-lysosomes to functional lysosomes. In this review, we will summarize progress in unveiling the molecular mechanisms underlying ALR and its potential pathophysiological roles.
Collapse
Affiliation(s)
- Yang Chen
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084,
China
| | - Li Yu
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084,
China
| |
Collapse
|
191
|
Guo F, Liu X, Cai H, Le W. Autophagy in neurodegenerative diseases: pathogenesis and therapy. Brain Pathol 2018; 28:3-13. [PMID: 28703923 PMCID: PMC5739982 DOI: 10.1111/bpa.12545] [Citation(s) in RCA: 273] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 06/30/2017] [Indexed: 12/12/2022] Open
Abstract
The most prevalent pathological features of many neurodegenerative diseases are the aggregation of misfolded proteins and the loss of certain neuronal populations. Autophagy, as major intracellular machinery for degrading aggregated proteins and damaged organelles, has been reported to be involved in the occurrence of pathological changes in many neurodegenerative disorders, including Alzheimer's disease, Parkinson's disease, Huntington's disease and amyotrophic lateral sclerosis. In this review, we summarize most recent research progress in this topic and provide a new perspective regarding autophagy regulation on the pathogenesis of neurodegenerative diseases. Finally, we discuss the signaling molecules in autophagy-related pathways as therapeutic targets for the treatment of these diseases.
Collapse
Affiliation(s)
- Fang Guo
- The Key Laboratory of Stem Cell Biology and Neurogenomic LaboratoryInstitute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences & Shanghai Jiao Tong University School of MedicineShanghai200025China
| | - Xinyao Liu
- Clinical Research Center on Neurological Diseasesthe First Affiliated Hospital, Dalian Medical UniversityDalian116011China
| | - Huaibin Cai
- Laboratory of NeurogeneticsNational Institute on Aging, National Institutes of HealthBethesdaMD
| | - Weidong Le
- The Key Laboratory of Stem Cell Biology and Neurogenomic LaboratoryInstitute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences & Shanghai Jiao Tong University School of MedicineShanghai200025China
- Clinical Research Center on Neurological Diseasesthe First Affiliated Hospital, Dalian Medical UniversityDalian116011China
- Collaborative Innovation Center for Brain Sciencethe First Affiliated Hospital, Dalian Medical UniversityDalian116011China
| |
Collapse
|
192
|
Mattera R, Park SY, De Pace R, Guardia CM, Bonifacino JS. AP-4 mediates export of ATG9A from the trans-Golgi network to promote autophagosome formation. Proc Natl Acad Sci U S A 2017; 114:E10697-E10706. [PMID: 29180427 PMCID: PMC5740629 DOI: 10.1073/pnas.1717327114] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
AP-4 is a member of the heterotetrameric adaptor protein (AP) complex family involved in protein sorting in the endomembrane system of eukaryotic cells. Interest in AP-4 has recently risen with the discovery that mutations in any of its four subunits cause a form of hereditary spastic paraplegia (HSP) with intellectual disability. The critical sorting events mediated by AP-4 and the pathogenesis of AP-4 deficiency, however, remain poorly understood. Here we report the identification of ATG9A, the only multispanning membrane component of the core autophagy machinery, as a specific AP-4 cargo. AP-4 promotes signal-mediated export of ATG9A from the trans-Golgi network to the peripheral cytoplasm, contributing to lipidation of the autophagy protein LC3B and maturation of preautophagosomal structures. These findings implicate AP-4 as a regulator of autophagy and altered autophagy as a possible defect in AP-4-deficient HSP.
Collapse
Affiliation(s)
- Rafael Mattera
- Cell Biology and Neurobiology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892
| | - Sang Yoon Park
- Cell Biology and Neurobiology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892
| | - Raffaella De Pace
- Cell Biology and Neurobiology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892
| | - Carlos M Guardia
- Cell Biology and Neurobiology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892
| | - Juan S Bonifacino
- Cell Biology and Neurobiology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
193
|
Abstract
Autophagy is an essential cellular process for bulk degradation of cytoplasmic components through the lysosome. Underlying this process is an intricate interaction between protein factors and the cell endomembrane system, leading to a gradual maturation of the autophagic membrane. This structure sequesters a portion of the cytoplasm by the formation of a double-membrane compartment called the autophagosome. The autophagosome then delivers the cargo to the lysosome to complete degradation. The molecular mechanism accounting for the generation of the autophagic membrane is a longstanding question. Here, a cell-free approach that has been established to understand the mechanism of early autophagic membrane generation is described. This system has provided insight into the membrane source of the autophagosome, the early protein-membrane associations, and the membrane remodeling that generates the autophagosomal precursors. The cell-free assay, in combination with other established approaches (e.g., cell imaging), will facilitate a deeper understanding of the mechanism of autophagy. © 2017 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Min Zhang
- State Key Laboratory of Membrane Biology, Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Dawei Liu
- Howard Hughes Medical Institute, Department of Molecular and Cell Biology, University of California, Berkeley
| | - Liang Ge
- State Key Laboratory of Membrane Biology, Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
194
|
Coppens I. How Toxoplasma and malaria parasites defy first, then exploit host autophagic and endocytic pathways for growth. Curr Opin Microbiol 2017; 40:32-39. [DOI: 10.1016/j.mib.2017.10.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 09/15/2017] [Accepted: 10/15/2017] [Indexed: 02/07/2023]
|
195
|
Fang EF, Palikaras K, Sun N, Fivenson EM, Spangler RD, Kerr JS, Cordonnier SA, Hou Y, Dombi E, Kassahun H, Tavernarakis N, Poulton J, Nilsen H, Bohr VA. In Vitro and In Vivo Detection of Mitophagy in Human Cells, C. Elegans, and Mice. J Vis Exp 2017. [PMID: 29286376 DOI: 10.3791/56301] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Mitochondria are the powerhouses of cells and produce cellular energy in the form of ATP. Mitochondrial dysfunction contributes to biological aging and a wide variety of disorders including metabolic diseases, premature aging syndromes, and neurodegenerative diseases such as Alzheimer's disease (AD) and Parkinson's disease (PD). Maintenance of mitochondrial health depends on mitochondrial biogenesis and the efficient clearance of dysfunctional mitochondria through mitophagy. Experimental methods to accurately detect autophagy/mitophagy, especially in animal models, have been challenging to develop. Recent progress towards the understanding of the molecular mechanisms of mitophagy has enabled the development of novel mitophagy detection techniques. Here, we introduce several versatile techniques to monitor mitophagy in human cells, Caenorhabditis elegans (e.g., Rosella and DCT-1/ LGG-1 strains), and mice (mt-Keima). A combination of these mitophagy detection techniques, including cross-species evaluation, will improve the accuracy of mitophagy measurements and lead to a better understanding of the role of mitophagy in health and disease.
Collapse
Affiliation(s)
- Evandro F Fang
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health; Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital; ;
| | - Konstantinos Palikaras
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology - Hellas
| | - Nuo Sun
- Center for Molecular Medicine, National Heart Lung and Blood Institute, National Institutes of Health
| | - Elayne M Fivenson
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health
| | - Ryan D Spangler
- Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health
| | - Jesse S Kerr
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health
| | - Stephanie A Cordonnier
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health
| | - Yujun Hou
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health
| | - Eszter Dombi
- Nuffield Department of Obstetrics and Gynaecology, University of Oxford
| | - Henok Kassahun
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology - Hellas; Department of Basic Sciences, Faculty of Medicine, University of Crete
| | - Joanna Poulton
- Nuffield Department of Obstetrics and Gynaecology, University of Oxford
| | - Hilde Nilsen
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital
| | - Vilhelm A Bohr
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health; Danish Center for Healthy Aging, University of Copenhagen;
| |
Collapse
|
196
|
Abstract
Soukup et al. (2016), in this issue of Neuron, and Murdoch et al. (2016), in Cell Reports, reveal an unexpected function for the endocytic protein endophilin in autophagosome formation at synapses: preventing neurodegeneration and ataxia.
Collapse
Affiliation(s)
- Marijn Kuijpers
- Leibniz-Institut für Molekulare Pharmakologie, 13125 Berlin, Germany
| | - Volker Haucke
- Leibniz-Institut für Molekulare Pharmakologie, 13125 Berlin, Germany.
| |
Collapse
|
197
|
Sun H, Liu Y, Zhang L, Shao X, Liu K, Ding Z, Liu X, Jiang C, Li H, Li H. Numb positively regulates autophagic flux via regulating lysosomal function. Biochem Biophys Res Commun 2017; 491:780-786. [PMID: 28720501 DOI: 10.1016/j.bbrc.2017.07.084] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 07/14/2017] [Indexed: 11/15/2022]
Abstract
Autophagy is a lysosome-dependent catabolic process involving in the degradation and recycling of unnecessary or damaged proteins and organelles. Emerging evidence indicates that autophagy dysfunction is closely related to various human diseases including cancer, aging, myopathies and neurodegenerative disorders. Here, using genetic knockdown, we uncover the role of Numb, an endocytic adaptor protein, in regulating the late steps of autophagy. We found that Numb depletion led to the accumulation of autophagic vacuole, as verified by RFP-LC3 staining combined with transmission electron microscopy. Further investigation indicated that Numb depletion impaired autophagic degradation through inhibiting the activities of lysosomal enzymes (Cathepsin D, β-glucuronidase and β-glucosidase). Moreover, Numb depletion induced elevation of lysosomal pH values and decrease of glycosylated lysosome-associated membrane proteins. We further observed that Rab7 activity was inhibited in Numb-depleted cells. Together, our findings revealed a novel function of Numb and its likely mechanism in regulation of autophagy events.
Collapse
Affiliation(s)
- Haiyan Sun
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, Guangdong Key Laboratory of Nanomedicine, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yi Liu
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, Guangdong Key Laboratory of Nanomedicine, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Lei Zhang
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, Guangdong Key Laboratory of Nanomedicine, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Ximing Shao
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, Guangdong Key Laboratory of Nanomedicine, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Ke Liu
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, Guangdong Key Laboratory of Nanomedicine, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Zhihao Ding
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, Guangdong Key Laboratory of Nanomedicine, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Xianming Liu
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, Guangdong Key Laboratory of Nanomedicine, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China; College of Life Science and Technology, Jinan University, 601 West Huangpu Blvd., Guangzhou 510632, China
| | - Changan Jiang
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, Guangdong Key Laboratory of Nanomedicine, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Huashun Li
- SARITEX Center for Stem Cell Engineering Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Advanced Institute of Translational Medicine, Shanghai 200123, China; ATCG Corporation, BioBay, Suzhou Industrial Park, Suzhou, Jiangsu 215123, China
| | - Hongchang Li
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, Guangdong Key Laboratory of Nanomedicine, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China.
| |
Collapse
|
198
|
Hammerling BC, Shires SE, Leon LJ, Cortez MQ, Gustafsson ÅB. Isolation of Rab5-positive endosomes reveals a new mitochondrial degradation pathway utilized by BNIP3 and Parkin. Small GTPases 2017; 11:69-76. [PMID: 28696827 DOI: 10.1080/21541248.2017.1342749] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Degradation of mitochondria is an important cellular quality control mechanism mediated by two distinct pathways: one involving Parkin-mediated ubiquitination and the other dependent on mitophagy receptors. It is known that mitochondria are degraded by the autophagy pathway; however, we recently reported that the small GTPase Rab5 and early endosomes also participate in Parkin-mediated mitochondrial clearance. Here, we have developed a protocol to isolate Rab5-positive vesicles from cells for proteomics analysis and provide additional data confirming that mitophagy regulators and mitochondrial proteins are present in these vesicles. We also demonstrate that the mitophagy receptor BNIP3 utilizes the Rab5-endosomal pathway to clear mitochondria in cells. These findings indicate that a redundancy exists in the downstream degradation pathways to ensure efficient mitochondrial clearance.
Collapse
Affiliation(s)
- Babette C Hammerling
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Sarah E Shires
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Leonardo J Leon
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Melissa Q Cortez
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Åsa B Gustafsson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
199
|
Kjos I, Borg Distefano M, Sætre F, Repnik U, Holland P, Jones AT, Engedal N, Simonsen A, Bakke O, Progida C. Rab7b modulates autophagic flux by interacting with Atg4B. EMBO Rep 2017; 18:1727-1739. [PMID: 28835545 PMCID: PMC5623852 DOI: 10.15252/embr.201744069] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 06/26/2017] [Accepted: 06/29/2017] [Indexed: 11/11/2022] Open
Abstract
Autophagy (macroautophagy) is a highly conserved eukaryotic degradation pathway in which cytosolic components and organelles are sequestered by specialized autophagic membranes and degraded through the lysosomal system. The autophagic pathway maintains basal cellular homeostasis and helps cells adapt during stress; thus, defects in autophagy can cause detrimental effects. It is therefore crucial that autophagy is properly regulated. In this study, we show that the cysteine protease Atg4B, a key enzyme in autophagy that cleaves LC3, is an interactor of the small GTPase Rab7b. Indeed, Atg4B interacts and co‐localizes with Rab7b on vesicles. Depletion of Rab7b increases autophagic flux as indicated by the increased size of autophagic structures as well as the magnitude of macroautophagic sequestration and degradation. Importantly, we demonstrate that Rab7b regulates LC3 processing by modulating Atg4B activity. Taken together, our findings reveal Rab7b as a novel negative regulator of autophagy through its interaction with Atg4B.
Collapse
Affiliation(s)
- Ingrid Kjos
- Department of Biosciences, University of Oslo, Oslo, Norway.,Centre for Immune Regulation, University of Oslo, Oslo, Norway
| | - Marita Borg Distefano
- Department of Biosciences, University of Oslo, Oslo, Norway.,Centre for Immune Regulation, University of Oslo, Oslo, Norway
| | - Frank Sætre
- Centre for Molecular Medicine Norway, University of Oslo, Oslo, Norway
| | - Urska Repnik
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Petter Holland
- Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Arwyn T Jones
- Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Wales, UK
| | - Nikolai Engedal
- Centre for Molecular Medicine Norway, University of Oslo, Oslo, Norway
| | - Anne Simonsen
- Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Oddmund Bakke
- Department of Biosciences, University of Oslo, Oslo, Norway .,Centre for Immune Regulation, University of Oslo, Oslo, Norway
| | - Cinzia Progida
- Department of Biosciences, University of Oslo, Oslo, Norway .,Centre for Immune Regulation, University of Oslo, Oslo, Norway
| |
Collapse
|
200
|
Assessment of Autophagy in Neurons and Brain Tissue. Cells 2017; 6:cells6030025. [PMID: 28832529 PMCID: PMC5617971 DOI: 10.3390/cells6030025] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 08/01/2017] [Accepted: 08/21/2017] [Indexed: 12/31/2022] Open
Abstract
Autophagy is a complex process that controls the transport of cytoplasmic components into lysosomes for degradation. This highly conserved proteolytic system involves dynamic and complex processes, using similar molecular elements and machinery from yeast to humans. Moreover, autophagic dysfunction may contribute to a broad spectrum of mammalian diseases. Indeed, in adult tissues, where the capacity for regeneration or cell division is low or absent (e.g., in the mammalian brain), the accumulation of proteins/peptides that would otherwise be recycled or destroyed may have pathological implications. Indeed, such changes are hallmarks of pathologies, like Alzheimer’s, Prion or Parkinson’s disease, known as proteinopathies. However, it is still unclear whether such dysfunction is a cause or an effect in these conditions. One advantage when analysing autophagy in the mammalian brain is that almost all the markers described in different cell lineages and systems appear to be present in the brain, and even in neurons. By contrast, the mixture of cell types present in the brain and the differentiation stage of such neurons, when compared with neurons in culture, make translating basic research to the clinic less straightforward. Thus, the purpose of this review is to describe and discuss the methods available to monitor autophagy in neurons and in the mammalian brain, a process that is not yet fully understood, focusing primarily on mammalian macroautophagy. We will describe some general features of neuronal autophagy that point to our focus on neuropathologies in which macroautophagy may be altered. Indeed, we centre this review around the hypothesis that enhanced autophagy may be able to provide therapeutic benefits in some brain pathologies, like Alzheimer’s disease, considering this pathology as one of the most prevalent proteinopathies.
Collapse
|